Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
2.
Clin Exp Immunol ; 212(1): 14-28, 2023 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-36879430

RESUMO

Allergen immunotherapy (AIT) is currently the only form of treatment that modifies allergic asthma. Pharmacotherapy alone seeks to control the symptoms of allergic asthma, allergic rhinitis, and other atopic conditions. In contrast, AIT can induce long-term physiological modifications through the immune system. AIT enables individuals to live improved lives many years after treatment ends, where they are desensitized to the allergen(s) used or no longer have significant allergic reactions upon allergen provocation. The leading forms of treatment with AIT involve injections of allergen extracts with increasing doses via the subcutaneous route or drops/tablets via the sublingual route for several years. Since the initial attempts at this treatment as early as 1911 by Leonard Noon, the mechanisms by which AIT operates remain unclear. This literature-based review provides the primary care practitioner with a current understanding of the mechanisms of AIT, including its treatment safety, protocols, and long-term efficacy. The primary mechanisms underlying AIT include changes in immunoglobulin classes (IgA, IgE, and IgG), immunosuppressive regulatory T-cell induction, helper T cell type 2 to helper T cell type 1 cell/cytokine profile shifts, decreased early-phase reaction activity and mediators, and increased production of IL-10, IL-35, TGF-ß, and IFN-γ. Using the databases PubMed and Embase, a selective literature search was conducted searching for English, full-text, reviews published between 2015 and 2022 using the keywords (with wildcards) "allerg*," "immunotherap*," "mechanis*," and "asthma." Among the cited references, additional references were identified using a manual search.


Assuntos
Asma , Rinite Alérgica , Humanos , Asma/terapia , Dessensibilização Imunológica/métodos , Alérgenos
3.
Tissue Eng Regen Med ; 20(3): 389-409, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36920675

RESUMO

Various immune cells participate in repair and regeneration following tissue injury or damage, orchestrating tissue inflammation and regeneration processes. A deeper understanding of the immune system's involvement in tissue repair and regeneration is critical for the development of successful reparatory and regenerative strategies. Here we review recent technologies that facilitate cell-based and biomaterial-based modulation of the immune systems for tissue repair and regeneration. First, we summarize the roles of various types of immune cells in tissue repair. Second, we review the principle, examples, and limitations of regulatory T (Treg) cell-based therapy, a representative cell-based immunotherapy. Finally, we discuss biomaterial-based immunotherapy strategies that aim to modulate immune cells using various biomaterials for tissue repair and regeneration.


Assuntos
Imunidade , Regeneração , Materiais Biocompatíveis , Imunomodulação
4.
Exp Dermatol ; 32(3): 256-263, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36458459

RESUMO

Immunosuppression in the tumour microenvironment (TME) attenuates antitumor immunity, consequently hindering protective immunosurveillance and preventing effective antitumor immunity induced by cancer immunotherapy. Multiple mechanisms including immune checkpoint molecules, such as CTLA-4, PD-1, and LAG-3, and immunosuppressive cells are involved in the immunosuppression in the TME. Regulatory T (Treg) cells, a population of immunosuppressive cells, play an important role in inhibiting antitumor immunity. Therefore, Treg cells in the TME correlate with an unfavourable prognosis in various cancer types. Thus, Treg cell is considered to become a promising target for cancer immunotherapy. Elucidating Treg cell functions in cancer patients is therefore crucial for developing optimal Treg cell-targeted immunotherapy. Here, we describe Treg cell functions and phenotypes in the TME from the perspective of Treg cell-targeted immunotherapy.


Assuntos
Neoplasias , Linfócitos T Reguladores , Humanos , Imunoterapia , Tolerância Imunológica , Terapia de Imunossupressão , Microambiente Tumoral
5.
Front Immunol ; 13: 966364, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36090981

RESUMO

FOXP3 is the lineage-defining transcription factor for Tregs, a cell type critical to immune tolerance, but the mechanisms that control FOXP3 expression in Tregs remain incompletely defined, particularly as it relates to signals downstream of TCR and CD28 signaling. Herein, we studied the role of IRF4 and BATF3, two transcription factors upregulated upon T cell activation, to the conversion of conventional CD4+ T cells to FOXP3+ T cells (iTregs) in vitro. We found that IRF4 must partner with BATF3 to bind to a regulatory region in the Foxp3 locus where they cooperatively repress FOXP3 expression and iTreg induction. In addition, we found that interactions of these transcription factors are necessary for glycolytic reprogramming of activated T cells that is antagonistic to FOXP3 expression and stability. As a result, Irf4 KO iTregs show increased demethylation of the critical CNS2 region in the Foxp3 locus. Together, our findings provide important insights how BATF3 and IRF4 interactions integrate activating signals to control CD4+ cell fate decisions and govern Foxp3 expression.


Assuntos
Fatores de Transcrição Forkhead , Fator de Transcrição AP-1 , Linfócitos T CD4-Positivos/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Linfócitos T Reguladores , Fator de Transcrição AP-1/metabolismo
6.
Front Immunol ; 13: 1068399, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36685593

RESUMO

T peripheral helper (Tph) cells, identified in the synovium of adults with seropositive rheumatoid arthritis, drive B cell maturation and antibody production in non-lymphoid tissues. We sought to determine if similarly dysregulated T cell-B cell interactions underlie another form of inflammatory arthritis, juvenile oligoarthritis (oligo JIA). Clonally expanded Tph cells able to promote B cell antibody production preferentially accumulated in the synovial fluid (SF) of oligo JIA patients with antinuclear antibodies (ANA) compared to autoantibody-negative patients. Single-cell transcriptomics enabled further definition of the Tph gene signature in inflamed tissues and showed that Tph cells from ANA-positive patients upregulated genes associated with B cell help to a greater extent than patients without autoantibodies. T cells that co-expressed regulatory T and B cell-help factors were identified. The phenotype of these Tph-like Treg cells suggests an ability to restrain T cell-B cell interactions in tissues. Our findings support the central role of disordered T cell-help to B cells in autoantibody-positive arthritides.


Assuntos
Artrite Juvenil , Artrite Reumatoide , Humanos , Autoanticorpos , Linfócitos T Auxiliares-Indutores , Linfócitos B
7.
Front Oncol ; 12: 1061789, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36703796

RESUMO

Triple negative breast cancer (TNBC) is an aggressive disease that is difficult to treat and portends a poor prognosis in many patients. Recent efforts to implement immune checkpoint inhibitors into the treatment landscape of TNBC have led to improved outcomes in a subset of patients both in the early stage and metastatic settings. However, a large portion of patients with TNBC remain resistant to immune checkpoint inhibitors and have limited treatment options beyond cytotoxic chemotherapy. The interplay between the anti-tumor immune response and tumor metabolism contributes to immunotherapy response in the preclinical setting, and likely in the clinical setting as well. Specifically, tumor glycolysis and lactate production influence the tumor immune microenvironment through creation of metabolic competition with infiltrating immune cells, which impacts response to immune checkpoint blockade. In this review, we will focus on how glucose metabolism within TNBC tumors influences the response to immune checkpoint blockade and potential ways of harnessing this information to improve clinical outcomes.

8.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-940520

RESUMO

ObjectiveTo observe the effect of asiaticoside (AC) on the expression of T helper 17 (Th17) cells and regulatory T (Treg) cells in DBA/1 mice with collagen-induced arthritis (CIA). MethodMale SPF DBA/1 mice were randomized into six groups according to body weight: control group, CIA group, methotrexate group (MTX group, ip, 0.5 mg·kg-1), and AC low-, medium-, and high-dose groups (ig, 5, 15, 45 mg·kg-1, respectively). Modeling was performed in rats other than the control group. To be specific, they were immunized with bovine type Ⅱ collagen and complete Freund's adjuvant on the first day and with bovine type Ⅱ collagen and incomplete Freund's adjuvant on the 21st day. Administration began on the day of the second immunization, once a day for 28 days. On the 49th day, related tissues were collected. Then, hematoxylin-eosin (HE) staining was performed to observe the pathological changes of the joints. Immunohistochemical method was used to detect the expression of interleukin-17 (IL-17) and forkhead box protein-3 (FoxP3), the markers of Th17 and Treg cells, respectively, immunofluorescence double staining the expression of IL-17 and FoxP3 in CD4+T cells of mouse joint tissue, and flow cytometry the proportions of Th17 and Treg cells in mouse lymph nodes. ResultCompared with the control group, CIA group demonstrated joint disorder, damage of articular cartilage and bone, severe bone erosion (P<0.01), increase in stained CD4 and IL-17 and the integral absorbance (IA) (P<0.01), decrease in stained FoxP3 and the IA (P<0.01), rise of Th17/Treg ratio (P<0.01), elevation of Th17 expression in mouse lymph nodes (P<0.01), and reduction in Treg expression (P<0.01). Compared with CIA group, MTX group and three AC groups showed normal joints, alleviated bone erosion and damage, intact and smooth joint surface, and decrease in stained IL-17 and IA (P<0.05, P<0.01), and MTX group and AC medium-dose and high-dose groups registered decrease in stained CD4 and IA (P<0.01) and reduction in Th17/Treg ratio (P<0.05, P<0.01). Moreover, AC medium-dose and high-dose groups showed rise in stained FoxP3 and IA (P<0.05, P<0.01). In the lymph nodes of mice, decrease in expression of Th17 cells (P<0.05, P<0.01) and the increase in expression of Treg cells (P<0.05, P<0.01) were observed in all the three AC group. ConclusionAC can regulate Th17/Treg balance by inhibiting the expression of Th17 cells and promoting the expression of Treg cells in CIA mice.

9.
Front Pharmacol ; 12: 726035, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34531749

RESUMO

Atopic dermatitis (AD), also known as atopic eczema, is one of the most common skin diseases and is characterized by allergic skin inflammation, redness, and itchiness and is associated with a hyperactivated type 2 immune response. The leading causes of AD include an imbalance in the immune system, genetic predisposition, or environmental factors, making the development of effective pharmacotherapies complex. Steroids are widely used to treat AD; however, they provide limited efficacy in the long term and can lead to adverse effects. Thus, novel treatments that offer durable efficacy and fewer side effects are urgently needed. Here, we investigated the therapeutic potential of Huangbai Liniment (HB), a traditional Chinese medicine, using an experimental AD mouse model, following our clinical observations of AD patients. In both AD patient and the mouse disease model, HB significantly improved the disease condition. Specifically, patients who received HB treatment on local skin lesions (3-4 times/day) showed improved resolution of inflammation. Using the 1-Chloro-2,4-dinitrobenzene (DNCB)-induced AD model in BALB/c mice, we observed that HB profoundly alleviated severe skin inflammation and relieved the itching. The dermatopathological results showed markedly reversed skin inflammation with decreased epidermal thickness and overall cellularity. Correspondingly, HB treatment largely decreased the mRNA expression of proinflammatory cytokines, including IL-1ß, TNF-α, IL-17, IL-4, and IL-13, associated with declined gene expression of IL-33, ST2, and GATA3, which are connected to the type 2 immune response. In addition, HB restored immune tolerance by promoting regulatory T (TREG) cells and inhibiting the generation of TH1, TH2, and TH17 cells in vitro and in the DNCB-induced AD mouse model. For the first time, we demonstrate that HB markedly mitigates skin inflammation in AD patients and the DNCB-induced AD mouse model by reinvigorating the T cell immune balance, shedding light on the future development and application of novel HB-based therapeutics for AD.

10.
Int Immunol ; 33(8): 435-446, 2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34235533

RESUMO

Regulatory T (Treg) cells, which are essential for maintaining self-tolerance, inhibit anti-tumor immunity, consequently hindering protective cancer immunosurveillance, and hampering effective anti-tumor immune responses in tumor-bearing hosts. Here, we show that depletion of Treg cells via targeting glycoprotein A repetitions predominant (GARP) induces effective anti-tumor immune responses. GARP was specifically expressed by highly suppressive Treg cells in the tumor microenvironment (TME) of multiple cancer types in humans. In the periphery, GARP was selectively induced in Treg cells, but not in effector T cells, by polyclonal stimulation. DS-1055a, a novel afucosylated anti-human GARP monoclonal antibody, efficiently depleted GARP+ Treg cells, leading to the activation of effector T cells. Moreover, DS-1055a decreased FoxP3+CD4+ T cells in the TME and exhibited remarkable anti-tumor activity in humanized mice bearing HT-29 tumors. We propose that DS-1055a is a new Treg-cell-targeted cancer immunotherapy agent with augmentation of anti-tumor immunity.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas de Membrana/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Humanos , Tolerância Imunológica/imunologia , Imunidade/imunologia , Imunoterapia/métodos , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Microambiente Tumoral/imunologia
11.
Front Oncol ; 11: 684098, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34141625

RESUMO

Immune checkpoint inhibitors (ICIs) have revolutionized the treatment paradigm for lung cancer in recent years. These strategies consist of neutralizing antibodies against negative regulators of immune function, most notably cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and PD-1 ligand 1 (PD-L1), thereby impeding the ability of tumor cells to escape immune surveillance. Though ICIs have proven a significant advance in lung cancer therapy, overall survival rates remain low, and lung cancer continues to be the leading cause of cancer-related death in the United States. It is therefore imperative to better understand the barriers to the efficacy of ICIs, particularly additional mechanisms of immunosuppression within the lung cancer microenvironment. Recent evidence suggests that regulatory T-lymphocytes (Tregs) serve as a central mediator of immune function in lung cancer, suppressing sterilizing immunity and contributing to the clinical failure of ICIs. Here, we provide a comprehensive summary of the roles of Tregs in lung cancer pathobiology and therapy, as well as the potential means through which these immunosuppressive mechanisms can be overcome.

12.
Front Immunol ; 11: 567981, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072108

RESUMO

Several recent studies have reported a key role for innate cell hyper-responsiveness in food allergy. This has predominantly been observed in early life, with evidence that innate immune function may return to baseline if food allergy resolves in later childhood. Hallmarks of hyper-responsiveness include increased circulating frequency of monocytes and altered innate cell cytokine responses to in vitro exposure with bacterial endotoxin. These features mirror the defining signatures of trained innate immunity, seen in other complex diseases. In this study, detailed immune cell and cytokine profiling was performed on peripheral blood mononuclear cells at baseline from 27 1 year old infants in the HealthNuts cohort (n = 16 egg allergic and n = 11 non-allergic healthy controls) and following monocyte stimulation. We show that egg allergic infants have increased frequency of circulating monocytes, reduced numbers of regulatory CD4 T cells and increased monocyte: CD4 T cell ratios relative to healthy controls. Monocytes from both egg allergic and non-allergic infants responded to endotoxin stimulation with rapid cytokine production and downregulation of the surface receptor CD16, however monocytes from egg allergic infants were hyper-responsive, producing significantly more inflammatory cytokines (TNFα, IL-6, IL-1ß, IL-8) and innate cell recruiting factors (MIP-1α) than healthy controls. This work indicates that monocytes of food allergic infants are programmed to a hyper-inflammatory phenotype and that the development of food allergy may be associated with trained immunity in early life.


Assuntos
Suscetibilidade a Doenças , Endotoxinas/efeitos adversos , Hipersensibilidade Alimentar/etiologia , Hipersensibilidade Alimentar/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Fatores Etários , Alérgenos/imunologia , Biomarcadores , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Citocinas/biossíntese , Hipersensibilidade a Ovo/diagnóstico , Hipersensibilidade a Ovo/etiologia , Hipersensibilidade a Ovo/metabolismo , Feminino , Hipersensibilidade Alimentar/diagnóstico , Humanos , Imunização , Imunoglobulina E/imunologia , Imunofenotipagem , Lactente , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino
13.
Front Immunol ; 11: 844, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32508814

RESUMO

Fusion proteins, which consist of factor VIII or factor IX and the transmucosal carrier cholera toxin subunit B, expressed in chloroplasts and bioencapsulated within plant cells, initiate tolerogenic immune responses in the intestine when administered orally. This approach induces regulatory T cells (Treg), which suppress inhibitory antibody formation directed at hemophilia proteins induced by intravenous replacement therapy in hemophilia A and B mice. Further analyses of Treg CD4+ lymphocyte sub-populations in hemophilia B mice reveal a marked increase in the frequency of CD4+CD25-FoxP3-LAP+ T cells (but not of CD4+CD25+FoxP3+ T cells) in the lamina propria of the small but not large intestine. The adoptive transfer of very small numbers of CD4+CD25-LAP+ Treg isolated from the spleen of tolerized mice was superior in suppression of antibodies directed against FIX when compared to CD4+CD25+ T cells. Thus, tolerance induction by oral delivery of antigens bioencapsulated in plant cells occurs via the unique immune system of the small intestine, and suppression of antibody formation is primarily carried out by induced latency-associated peptide (LAP) expressing Treg that likely migrate to the spleen. Tolerogenic antigen presentation in the small intestine requires partial enzymatic degradation of plant cell wall by commensal bacteria in order to release the antigen. Microbiome analysis of hemophilia B mice showed marked differences between small and large intestine. Remarkably, bacterial species known to produce a broad spectrum of enzymes involved in degradation of plant cell wall components were found in the small intestine, in particular in the duodenum. These were highly distinct from populations of cell wall degrading bacteria found in the large intestine. Therefore, FIX antigen presentation and Treg induction by the immune system of the small intestine relies on activity of a distinct microbiome that can potentially be augmented to further enhance this approach.


Assuntos
Toxina da Cólera/imunologia , Fator IX/imunologia , Microbioma Gastrointestinal/imunologia , Hemofilia B/imunologia , Hemofilia B/microbiologia , Tolerância Imunológica/imunologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Células Vegetais/metabolismo , Administração Oral , Transferência Adotiva/métodos , Animais , Apresentação de Antígeno , Antígenos/genética , Antígenos/imunologia , Toxina da Cólera/genética , Modelos Animais de Doenças , Fator IX/genética , Deleção de Genes , Genoma de Cloroplastos , Lactuca/genética , Masculino , Camundongos , Camundongos Transgênicos , Plantas Geneticamente Modificadas , Linfócitos T Reguladores/imunologia
14.
Front Immunol ; 11: 773, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32425946

RESUMO

The thymus is the central lymphoid organ for T cell development, a cradle of T cells, and for central tolerance establishment, an educator of T cells, maintaining homeostatic cellular immunity. T cell immunity is critical to control cancer occurrence, relapse, and antitumor immunity. Evidence on how aberrant thymic function influences cancer remains largely insufficient, however, there has been recent progress. For example, the involuted thymus results in reduced output of naïve T cells and a restricted T cell receptor (TCR) repertoire, inducing immunosenescence and potentially dampening immune surveillance of neoplasia. In addition, the involuted thymus relatively enhances regulatory T (Treg) cell generation. This coupled with age-related accumulation of Treg cells in the periphery, potentially provides a supportive microenvironment for tumors to escape T cell-mediated antitumor responses. Furthermore, acute thymic involution from chemotherapy can create a tumor reservoir, resulting from an inflammatory microenvironment in the thymus, which is suitable for disseminated tumor cells to hide, survive chemotherapy, and become dormant. This may eventually result in cancer metastatic relapse. On the other hand, if thymic involution is wisely taken advantage of, it may be potentially beneficial to antitumor immunity, since the involuted thymus increases output of self-reactive T cells, which may recognize certain tumor-associated self-antigens and enhance antitumor immunity, as demonstrated through depletion of autoimmune regulator (AIRE) gene in the thymus. Herein, we briefly review recent research progression regarding how altered thymic function modifies T cell immunity against tumors.


Assuntos
Neoplasias/imunologia , Timo/fisiologia , Antígenos de Neoplasias/imunologia , Humanos , Metástase Neoplásica , Neoplasias/etiologia , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Microambiente Tumoral
15.
Front Immunol ; 11: 444, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32296416

RESUMO

Foxp3-expressing regulatory T (Treg) cells are critical mediators of immunological tolerance to both self and microbial antigens. Tregs activate context-dependent transcriptional programs to adapt effector function to specific tissues; however, the factors controlling tissue-specific gene expression in Tregs remain unclear. Here, we find that the AP-1 transcription factor JunB regulates the intestinal adaptation of Tregs by controlling select gene expression programs in multiple Treg subsets. Treg-specific ablation of JunB results in immune dysregulation characterized by enhanced colonic T helper cell accumulation and cytokine production. However, in contrast to its classical binding-partner BATF, JunB is dispensable for maintenance of effector Tregs as well as most specialized Treg subsets. In the Peyer's patches, JunB activates a transcriptional program facilitating the maintenance of CD25- Tregs, leading to the complete loss of T follicular regulatory cells in the absence of JunB. This defect is compounded by loss of a separate effector program found in both major colonic Treg subsets that includes the cytolytic effector molecule granzyme B. Therefore, JunB is an essential regulator of intestinal Treg effector function through pleiotropic effects on gene expression.


Assuntos
Mucosa Intestinal/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Granzimas/metabolismo , Homeostase , Tolerância Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição AP-1/metabolismo
16.
Immun Ageing ; 17: 2, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31988649

RESUMO

Immune system aging is characterized by the paradox of immunosenescence (insufficiency) and inflammaging (over-reaction), which incorporate two sides of the same coin, resulting in immune disorder. Immunosenescence refers to disruption in the structural architecture of immune organs and dysfunction in immune responses, resulting from both aged innate and adaptive immunity. Inflammaging, described as a chronic, sterile, systemic inflammatory condition associated with advanced age, is mainly attributed to somatic cellular senescence-associated secretory phenotype (SASP) and age-related autoimmune predisposition. However, the inability to reduce senescent somatic cells (SSCs), because of immunosenescence, exacerbates inflammaging. Age-related adaptive immune system deviations, particularly altered T cell function, are derived from age-related thymic atrophy or involution, a hallmark of thymic aging. Recently, there have been major developments in understanding how age-related thymic involution contributes to inflammaging and immunosenescence at the cellular and molecular levels, including genetic and epigenetic regulation, as well as developments of many potential rejuvenation strategies. Herein, we discuss the research progress uncovering how age-related thymic involution contributes to immunosenescence and inflammaging, as well as their intersection. We also describe how T cell adaptive immunity mediates inflammaging and plays a crucial role in the progression of age-related neurological and cardiovascular diseases, as well as cancer. We then briefly outline the underlying cellular and molecular mechanisms of age-related thymic involution, and finally summarize potential rejuvenation strategies to restore aged thymic function.

17.
Front Immunol ; 10: 1954, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31475013

RESUMO

There is increasing evidence that T lymphocytes play a key role in controlling endogenous regeneration. Regeneration appears to be impaired in case of local accumulation of CD8+ effector T cells (TEFF), impairing endogenous regeneration by increasing a primary "useful" inflammation toward a damaging level. Thus, rescuing regeneration by regulating the heightened pro-inflammatory reaction employing regulatory CD4+ T (TReg) cells could represent an immunomodulatory option to enhance healing. Hypothesis was that CD4+ TReg might counteract undesired effects of CD8+ TEFF. Using adoptive TReg transfer, bone healing was consistently improved in mice possessing an inexperienced immune system with low amounts of CD8+ TEFF. In contrast, mice with an experienced immune system (high amounts of CD8+ TEFF) showed heterogeneous bone repair with regeneration being dependent upon the individual TEFF/TReg ratio. Thus, the healing outcome can only be improved by an adoptive TReg therapy, if an unfavorable TEFF/TReg ratio can be reshaped; if the individual CD8+ TEFF percentage, which is dependent on the individual immune experience can be changed toward a favorable ratio by the TReg transfer. Remarkably, also in patients with impaired fracture healing the TEFF/TReg ratio was higher compared to uneventful healers, validating our finding in the mouse osteotomy model. Our data demonstrate for the first time the key-role of a balanced TEFF/TReg response following injury needed to reach successful regeneration using bone as a model system. Considering this strategy, novel opportunities for immunotherapy in patients, which are at risk for impaired healing by targeting TEFF cells and supporting TReg cells to enhance healing are possible.


Assuntos
Desenvolvimento Ósseo/imunologia , Regeneração Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunomodulação/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva/métodos , Animais , Biomarcadores/sangue , Osso e Ossos/imunologia , Feminino , Fraturas Ósseas/terapia , Humanos , Imunoterapia/métodos , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Linfócitos T Reguladores/transplante
18.
Front Immunol ; 10: 1860, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31456800

RESUMO

Lipid metabolism plays a key role in many cellular processes. We show here that regulatory T cells have enhanced lipid storage within subcellular lipid droplets (LD). They also express elevated amounts of both isoforms of diacylglycerol acyl transferase (DGAT1 & 2), enzymes required for the terminal step of triacylglycerol synthesis. In regulatory T-cells (Tregs), the conversion of diacylglycerols to triacylglycerols serves two additional purposes other than lipid storage. First, we demonstrate that it protects T cells from the toxic effects of saturated long chain fatty acids. Second, we show that Triglyceride formation is essential for limiting activation of protein kinase C via free diacyl glycerol moieties. Inhibition of DGAT1 resulted in elevated active PKC and nuclear NFKB, as well as impaired Foxp3 induction in response to TGFß. Thus, Tregs utilize a positive feedback mechanism to promote sustained expression of Foxp3 associated with control of LD formation.


Assuntos
Fatores de Transcrição Forkhead/genética , Linfócitos T Reguladores/metabolismo , Triglicerídeos/metabolismo , Animais , Antígenos CD2/genética , Antígeno CD52/genética , Linhagem Celular , Diacilglicerol O-Aciltransferase/metabolismo , Ácidos Graxos/metabolismo , Feminino , Fatores de Transcrição Forkhead/biossíntese , Humanos , Gotículas Lipídicas/metabolismo , Metaboloma , Camundongos , Proteína Quinase C/metabolismo , Linfócitos T Reguladores/enzimologia , Linfócitos T Reguladores/imunologia
19.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-793192

RESUMO

@#Objective: To study the effect of anti-aging Klotho protein on immune escape mediated by regulatory T cells (Treg)/helper T cells 17 (TH17) in mice bearing cervical cancer and its mechanism. Methods: The model of cervical cancer-bearing mice were established, and the control group (normal mice), model group (cervical cancer-bearing mice model), and Klotho treatment group (cervical cancer-bearing mice treated with Klotho protein, 200 ng/d) were set up. The weight of cervical cancer tumors in mice of each group was weighed at 7 and 14 days after treatment respectively, PBMCs were separated at the same time. Flow cytometry was used to detect the changes of T lymphocyte function and the proportion of Treg and TH17 cells in mice. qPCR was used to detect the expressions of Foxp3 and RORγt, the key transcription factors of Treg/TH17 cells, in PBMCs of mice in each group. The changes of IL-17, IL-6, IL10, TGF-β and IL-23 in PBMCs were detected by ELISA. The protein expressions of Klotho, TGF-β, Foxp3 and RORγt in PBMCs of mice were detected by WB assay. Results: On the 14th day, the tumor inhibition rate of the cervical cancer-bearing mice in the Klotho group was significantly higher than that in the Model group [(52.16±8.25)% vs (23.33±6.29)% the model group to be supplemented, P< 0.05). Compared with the Control group, the ratios of Treg and TH17 cells in the lymphocytes of the tumor-bearing mice significantly increased (all P<0.05), the ratios of total T lymphocytes (CD3+), auxiliary/induced T lymphocytes (CD3+CD4+) and immune index (CD3+CD4+/CD3+CD8+ cells) decreased significantly (all P<0.05); in addition, the mRNAexpressions of Foxp3 and RORγt genes, cytokines of IL-17, IL-6, IL-10, TGF-β and IL-23, as well as protein expressions of TGF-β1, Foxp3 and RORγt increased significantly (all P <0.05), while the level of Klotho protein significantly decreased in Model group (P<0.05). Compared with the Model group, the above indicators showed opposite changes in Klotho group (P<0.05), but there was no significant difference with the Control group (all P> 0.05). Conclusion: Klotho protein may inhibit Treg/TH17 cell-mediated immune evasion in cervical cancer-bearing mice by inhibiting TGF-β1/Foxp3/RORγt signaling pathway and exert anti-tumor effect.

20.
J Allergy Clin Immunol ; 142(6): 1679-1695, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30527062

RESUMO

Monogenic diseases of the immune system, also known as inborn errors of immunity, are caused by single-gene mutations resulting in immune deficiency and dysregulation. More than 350 diseases have been described to date, and the number is rapidly expanding, with increasing availability of next-generation sequencing facilitating the diagnosis. The spectrum of immune dysregulation is wide, encompassing deficiencies in humoral, cellular, innate, and adaptive immunity; phagocytosis; and the complement system, which lead to autoinflammation and autoimmunity. Multiorgan autoimmunity is a dominant symptom when genetic mutations lead to defects in molecules essential for the development, survival, and/or function of regulatory T (Treg) cells. Studies of "Tregopathies" are providing critical mechanistic information on Treg cell biology, the role of Treg cell-associated molecules, and regulation of peripheral tolerance in human subjects. The pathogenic immune networks underlying these diseases need to be dissected to apply and develop immunomodulatory treatments and design curative treatments using cell and gene therapy. Here we review the pathogenetic mechanisms, clinical presentation, diagnosis, and current and future treatments of major known Tregopathies caused by mutations in FOXP3, CD25, cytotoxic T lymphocyte-associated antigen 4 (CTLA4), LPS-responsive and beige-like anchor protein (LRBA), and BTB domain and CNC homolog 2 (BACH2) and gain-of-function mutations in signal transducer and activator of transcription 3 (STAT3). We also discuss deficiencies in genes encoding STAT5b and IL-10 or IL-10 receptor as potential Tregopathies.


Assuntos
Síndromes de Imunodeficiência/imunologia , Linfócitos T Reguladores/imunologia , Animais , Fatores de Transcrição Forkhead/imunologia , Humanos , Síndromes de Imunodeficiência/diagnóstico , Interleucina-10/imunologia , Receptores de Interleucina-10/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...